AU4120200A - Inhibition of viral infection using monovalent antigen-binding proteins - Google Patents

Inhibition of viral infection using monovalent antigen-binding proteins Download PDF

Info

Publication number
AU4120200A
AU4120200A AU41202/00A AU4120200A AU4120200A AU 4120200 A AU4120200 A AU 4120200A AU 41202/00 A AU41202/00 A AU 41202/00A AU 4120200 A AU4120200 A AU 4120200A AU 4120200 A AU4120200 A AU 4120200A
Authority
AU
Australia
Prior art keywords
virus
antigen binding
phage
binding
vhh
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU41202/00A
Other versions
AU776824B2 (en
Inventor
Sandra Bezemer
Johannes Josef Wilhelmus De Haard
Leon Gerardus Joseph Frenken
Adrianus Marinus Ledeboer
Cornelis Theodorus Verrips
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Unilever PLC
Original Assignee
Unilever PLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Unilever PLC filed Critical Unilever PLC
Publication of AU4120200A publication Critical patent/AU4120200A/en
Application granted granted Critical
Publication of AU776824B2 publication Critical patent/AU776824B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Cosmetics (AREA)

Description

WO 00/65057 PCT/EPOO/03717 INHIBITION OF VIRAL INFECTION USING MONOVALENT ANTIGEN-BINDING PROTEINS FIELD OF THE INVENTION 5 The present invention relates to the use of antigen binding proteins in a method of inhibiting the infectivity of viruses or other infectious agents, products and compositions comprising such proteins and methods for identifying and/or selecting antigen binding proteins 10 capable of exhibiting such activity. In particular, the invention relates to a method for inhibiting viral infection using a monovalent antigen binding protein comprising a variable domain of a heavy chain derived from an immunoglobulin devoid of light chains, capable of binding to a virus. 15 BACKGROUND OF THE INVENTION Antibodies are protein molecules belonging to a group of immunoglobulins generated by the immune system in response to an 20 antigen. The structure of most antibody molecules is based on a unit comprising four polypeptides, two identical heavy chains and two identical light chains, which are covalently linked together by disulphide bonds. Each of these chains is folded in discrete domains. The C-terminal regions of both heavy and light chains are conserved in 25 sequence and are called the constant regions, comprising one or more so-called C-domains. The N-terminal regions of the heavy and light chains, also known as V-domains, are variable in sequence and determine the specificity of the antibody. The regions in the variable domains of the light and heavy chains (VL and VH respectively) 30 responsible for antigen binding activity are known as the hypervariable or complementarity determining regions (CDR) . Inmunoglobulins capable of exhibiting the functional properties of the four-chain immunoglobulins described above but which comprise two 35 heavy polypeptide chains and which furthermore are devoid of light polypeptide chains have been described (WO 94/04678, Casterman et al, 1994). Fragments corresponding to isolated VH domains (hereinafter VHH) are also disclosed. Methods for the preparation of such WO 00/65057 PCT/EPOO/03717 -2 antibodies or fragments thereof on a large scale comprising transforming a mould or yeast with an expressible DNA sequence encoding the antibody or fragment are described in patent application WO 94/25591 (Unilever). 5 The immunoglobulins described in WO 94/04678, which may be isolated from the serum of Camelids, do not rely upon the association of heavy and light chain variable domains for the formation of the antigen binding site but instead the heavy polypeptide chains alone naturally 10 form the complete antigen binding site. These immunoglobulins, hereinafter referred to as "heavy-chain immunoglobulins" are thus quite distinct from the heavy chains obtained by the degradation of common (four-chain) immunoglobulins or by direct cloning which contribute part only of the antigen-binding site and require a light 15 chain partner for antigen-binding, thus forming a complete antigen binding site. Antibodies or fragments thereof, have found application in a variety of uses where the specific nature of the antibody-antigen interaction 20 can be used to advantage. These include such uses as diagnosis, therapy, immunoassays and purification processes. The use of antibodies, or fragments thereof, in inhibiting viral infection has received attention, for instance during active immunisation with inactivated virus preparations or viral antigens produced in 25 recombinant cells or during passive immunisation by the administration of neutralising antibodies. It has been reported in the literature that monovalent Fab antibody fragments can neutralise viruses. Cheung et al (1992), Journal of 30 Virology, 66, 6714-6720, describe the production of the Fab domain of a rabies virus-neutralising antibody MAb-57 and further demonstrate that this monovalent fragment itself has virus-neutralising activity. Other publications also report the capability of human Fab monovalent antibody fragments to neutralise or inhibit viral activity (see for 35 example, Williamson et al (1993), Proc. Natl. Acad. Sci. USA, 90, 4141-4145). Such methods are not suitable for wide scale industrial application as the cost of producing such classical antibody fragments renders the processes economically unfeasible.
WO 00/65057 PCT/EP0O/03717 -3 An alternative approach to inhibiting viral replication using antibodies which has been described in the literature is to select antibodies to target enzymes produced by the virus. Martin et al, Protein Engineering, 10(5), 607-614 (1997) describes the use of a 5 camelisedf VH antibody fragment to inhibit hepatitis C virus NS3 protease, thereby preventing cleavage of the viral poly-protein precursor. Another industrial application in which economically viable solutions 10 to the problem of viral infection are sought is the field of fermentation processing, particularly food processing. Lactic acid bacteria (LAB: Lactococci and Lactobacilli) play an important role in food fermentation processes such as the production 15 of cheese or yoghurt. Often such fermentations are hampered by the sensitivity of the bacteria towards viruses, known as bacteriophage, which build up in these, often not aseptically performed, processes. A phage infection causes the LAB cells to lyse; during prolonged fermentations phage resistant cell populations can evolve, but this 20 delay affects the production capacity severely, and the disturbed process yields a product of low quality. Sometimes the process has to be stopped prematurely, with complete loss of the batch of milk. To date, the phage problem has mainly been approached by taking 25 special precautions with respect to hygiene at the production facility, but this causes additional time delays. Another solution which has been proposed is the use of resistant LAB strains, but the regular appearance of adapted forms of bacteriophage forces the strains used to be changed from time to time in a procedure known as 30 culture rotation. This has the disadvantage of requiring labour intensive monitoring of the production facilities and medium for the presence of phage and requires the availability of several sets of cultures with the same functional attributes, differing only in phage sensitivity. There therefore remains considerable commercial interest 35 in the further development of methods for combating LAB phage infection.
WO 00/65057 PCT/EPOO/03717 -4 One method, proposed by Geller et al (1998), J. Dairy Sci., 81, 895 900, involves the use of colostrum from cows immunized with lactococcal phage as a source of phage-neutralising (polyclonal) antibodies to prevent lytic infection of Lactococcus lactis in 5 fermentations of phage-contaminated milk. This method does not provide a commercially viable soluzon to the problem, however. Not only is it extremely economically unattractive to produce antibodies in this way but furthermore, the addition of colostrum to milk does not have regulatory approval. 10 An alternative approach, which makes use of multivalent, multispecific antigen binding proteins comprising a polypeptide comprising in series two or more single domain binding units, preferably variable domains of a heavy chain derived from an immunoglobulin naturally devoid of 15 light chains, to reduce the infectivity of LAB phages by cross-linking or agglutination is exemplified in the Applicant's co-pending patent application number PCT/EP98/06991, filed 26th October 1998. There remains a continuing need for the development of improved 20 methods of inhibiting or neutralising viral infection. In particular, there remains continuing interest in development of methods which can be applied economically on a scale appropriate for industrial use. SUMMARY OF THE INVENTION 25 Accordingly, the invention provides in one aspect a method of inhibiting viral infection using a monovalent antigen binding protein comprising a single variable domain binding unit, or a functional equivalent thereof, capable of binding to a virus. 30 In another aspect the invention provides the use of a monovalent antigen binding protein comprising a single variable domain binding unit or a functional equivalent thereof capable of binding to a virus in inhibiting viral infection. 35 The invention also provides the use of a monovalent antigen binding protein comprising a single variable domain binding unit or a functional equivalent thereof capable of binding to a virus in the WO 00/65057 PCT/EPOO/03717 preparation of a medicament for inhibiting viral infection. Also provided are monovalent antigen binding proteins comprising a single variable domain binding unit capable of binding to a virus, 5 nucleotide sequences encoding such proteins, cloning and expression vectors comprising such nucleotide sequences, host cells transformed with vectors comprising such nucleotide sequences, and food, cosmetic and pharmaceutical products comprising such proteins. 10 In a further aspect, the invention provides a method for selecting an antigen binding protein capable of inhibiting viral infection of a host cell comprising the steps of: i) complexing an antigen binding protein with a target virus, 15 ii) exposing the antigen binding protein-virus complex of step (i) to an excess of host cells, iii) removing the host cells and any associated antigen binding 20 protein-virus complex, iv) capturing antigen binding protein-virus complex not taken up by the host cells in step (ii) with virus specific ligands to separate virus specific antigen binding proteins from non 25 binding proteins. The invention also provides a method for identifying an antigen binding protein capable of inhibiting bacteriophage infection of a lactic acid bacterial cell host comprising the steps of: 30 i) culturing of bacterial host cells in the presence of antigen binding protein and bacteriophage, ii) assaying said culture for active cell growth manifest in a 35 change in pH of the culture growth medium. As used herein, a single variable domain binding unit means an immunoglobulin variable domain or a functional equivalent thereof WO 00/65057 PCT/EPOO/03717 -6 which forms a complete antigen binding site. This may be derived from natural sources or synthetically produced. The terms '-imunoglobulin' and 'antibody' are used synonymously throughout the specification, unless indicated otherwise. 5 A 'functional equivalent' of an imunoglobulin variable domain is any homogolous protein molecule which has similar binding specificity. A functional equivalent may be characterised by an insertion, deletion or substitution of one or more amino acid residues in the sequence of 10 the immunoglobulin variable domain. Suitably, the amino acid sequence of the functional equivalent has at least 60% similarity, preferably at least 80%, more preferably at least 90% similarity to the amino acid sequence of the immunoglobulin variable domain. 15 Inhibition of viral infection includes but is not limited to inhibition of infection by blocking essential sites on the viral particle, such as the receptor binding protein of the virus by which the virus attaches to the host cell during the first step of infection. Inhibition may be total or partial. The terms 'inhibit' 20 and 'neutralise' are used synonomously herein. The term 'virus' includes within its scope viruses, which infect bacterial host cells, known as bacteriophages. Binding to a virus includes binding to one or more molecules located at the surface of 25 the virus particle. The present invention may be more fully understood with reference to the following description when read together with the accompanying drawings in which: 30 Figure 1 shows the efficiency of the selected monovalent VHH fragment VHH#1 in neutralising Lactococcus lactis bacteriophage P2 as measured by plaque titration. 35 Figure 2 shows the prevention of phage infection by VHH fragment VHH#1 in an acidification experiment with a small scale culture of milk.
WO 00/65057 PCT/EPOO/03717 -7 Figure 3 shows the determination of the valency of the antibody fragments used in this study with an ELISA based method. DETAILED DESCRIPTION OF THE INVENTION 5 The invention is based on the finding that a monovalent antigen binding protein comprising a single variable domain binding unit capable of binding to a virus can advantageously be used to inhibit infection of a host cell by the virus. High production levels and 10 ease of downstream processing permits such antigen binding proteins to be applied in industrial processes and products economically and efficiently. As discussed above, antibody based methods for inhibiting viral 15 infection which have previously been described in the literature have relied either on cross-linking mediated via multivalent constructs or have made use of larger fragments derived from 'classical' antibodies, such as Fab fragments, to block the receptor binding protein of the virus, hence inhibiting its ability to infect the host cell. 20 Surprisingly, the present inventors have found that much smaller monovalent antigen binding proteins comprising a single variable domain are effective in inhibiting viral infection. This would not have been predicted from the prior art teaching as the smaller size of 25 these proteins might have been expected to have rendered them less effective in hindering binding of the virus to the host cell. Moreover, it would be expected that multivalent antigen binding proteins would be more effective due to the agglutination of infectious particles. To date, only the recognition of simple protein 30 antigens with such binding proteins has been reported. There has been no suggestion that complex systems such as viruses could be detected and inhibited using single domain binding units. The invention is applicable to the use of any immunoglobulin variable 35 domain, which forms a complete antigen binding site. The immunoglobulin may be derived from natural sources or synthetically produced. Preferably, the invention relates to the use of heavy chain variable domains derived from an immunoglobulin devoid of light WO 00/65057 PCT/EPOO/03717 chains, most suitably from an irmunoglobulin naturally devoid of light chains such as are obtainable from lymphoid cells, especially peripheral blood lymphocytes, bone marrow cells or spleen cells derived from Camelids as described in WO 94/04678 (Casterman et al) 5 discussed above. It will be appreciated that heavy chain variable domains derived from other immunoglobulins modified ('camelised') to enable them to function as monovalent binding domains in the same way as the heavy 10 chain variable domains derived from Camelids may also suitably be used according to the invention. An advantage of using single domain binding units which are heavy chain variable domains derived from Camelids is that they can readily 15 and conveniently be produced economically on a large scale, for example using a transformed lower eukaryotic host as described in WO 94/25591 (Unilever). A major advantage of the described production system is the low degree of impurities present in the secreted fraction, thereby enabling simple down stream processing procedures 20 for purification. A further advantage, particularly when applications in food processing are contemplated, is that such heavy chain variable domains are extremely heat stable, allowing pasteurisation or other heat treatments without loss of antigen binding capacity. 25 The invention is applicable to both prokaryotic and eukaryotic host cells. For therapy of humans or animals, target viruses of interest include pathogenic viruses such as those which belong to the family of Human Immunodeficiency Viruses. Other viral infections to which the invention is applicable include such food-born viruses as Hepatitis 30 viruses (especially Hepatitis A virus), Rotavirus and the small round Structured viruses (SRSV), such as Norwalk virus (see Food Science and Technology Today, II(1), 49-51, 1997). In the area of production crops, viruses pathogenic to plants such as Citrus tristeza virus (CTV), Tobacco mosaic virus (TMV), Potato virus Y (PVY), Lettuce 35 necrotic yellows virus (LNYV), Tomato spotted wilt virus (TSWV), Clover wound tumour virus (CWTV), Cauliflower mosaic virus (CaMV), Cowpea mosaic virus (CPMV), Soil-borne wheat furiovirus (SBWMV), Wheat yellow mosaic bymovirus (WYMV) and Wheat spindle streak mosaic virus WO 00/65057 PCT/EPOO/03717 -9 (WSSMV) are important targets for neutralisation or inhibition. Single domain binding units such as heavy chain variable domains recognising plant viruses can be cloned and expressed in plants using 5 methods equivalent to conventional cloning and expression of (modified) viral proteins, to protect these plants viruses. By using the appropriate targeting signals known in the art, the expression and translocation of the VHH's can be regulated in such a way that organelle or in the extracellular matrix. 10 The invention is of particular use in industrial fermentation processes, for example neutralising or inhibiting the infectivity of lactococcal bacteriophage, thereby preventing lytic infection of Lactococcus lactis. By inhibiting the infectivity of lactococcal 15 bacteriophages, the invention affords the possibility of avoiding having to adopt the various cost affecting measures described above. The antigen binding proteins can be used in a cleaning product, which removes phage present in the production system. Alternatively, they can be added to milk contaminated with bacteriophage, as is shown in 20 Example 3 below, which can be fermented to a high quality product without any delay in the production. The standard addition of such antigen binding proteins to milk would be one way in which it might be possible to abolish monitoring for the presence of phage. 25 Lactic acid bacteria play an important part in fermentation of many other food products in addition to dairy products. It will be appreciated that the invention is not restricted to use in inhibiting LAB phage infection in dairy fermentation processes but extends also to use in any process which makes use of lactic acid bacteria 30 fermentation. Suitable fermented food products and the associated lactic acid bacteria are listed in tables la-lb below (see Biotechnology, Vol 5, Chapter 1-8).
WO 00/65057 PCT/EPOO/03717 - 10 Table la Main functional lactic acid bacteria in European Fermented Foods PRODUCT NAME SUBSTRATE MAIN LACTIC ACID BACTERIA Baked Goods Wheat La:obacillus clantarum acidophilus delbrueckii brevis buchneri fermentum s.francisco Wine & Brandy Grapes Leuconostoe gracile oenos Lactobacillus plantarum case fructivorans hil gardii brevis Pediococcus cerevisiae Cheese & Dairy Milk Brevibacterium linens Products Lactococcus lactis cremoris Lactobacillus casei helveticus bulgaricus plantarum Leuconostoc cremoris Pediococcus acidilactici pentosaceus Sreptococcue thermophilus Enterococcus faecium Fermented Cabbage & Lactobacillus brevis vegetables/fruits Cucumbers plantarum Leuconostoc mesenteroides Pediococcus cerevisiae olives Lactobacillus plantarum paracasei brevis delbrueckii Streptococcus sp. Pediococcus sp. Leuconostoc sp. Sausages Meat Lactobacullus curvatus lactis plan tarum sake Pediococcus acidilactici pentocaceus Micrococcus caseolyticus WO 00/65057 PCT/EPOO/03717 - 11 Table lb Main functional lactic acid bacteria in Indigenous Fermented Foods PRODUCT NAME SUBSTRATES MAIN LACTIC ACID BACTERIA (Country) Banku (Ghana) Maize, cassava Lactic acid bacteria Burukutu (Nigeria) Sorghum, cassava Lactic acid bacteria Busa (Egypt) Rice, millet Lactobacillus sp. Dawadawa (Nigeria) Locust bean Lactic acid bacteria Dosai (India) Black gram and rice Lauconostoc mesenteroides Hamanatto (Japan) Whole soybean, Streptococcus sp. wheat flour Pediococcus sp. Idli (India) Rice, black gram Leuconostoc mesenteroides Kecap (Indonesia) Soybean, wheat Lactobacillus sp. Kimchi (Korea) Vegetables Lactic acid bacteria (seafood, nuts) Kishk (Egypt) Wheat, milk Lactic acid bacteria Mshewu (S. Africa) Maize Lactobacillus delbrueckii Miso (China, Japan) Rice and soybean Lactobacillus sp. Rice and cereals Lactobacillus sp. Ogi (Nigeria) Maize Lactic acid bacteria Puto (Philippines) Rice Lactic acid bacteria Sorghum beer Sorghum, maize Lactic acid bacteria (S. Africa) Soybean milk (Asia) Soybean Lactic acid bacteria Soy sauce (Asia) Soybean and wheat Lactobacillus sp. Pediococcus sp. Tarhana (Turkey) Wheat and milk Lactic acid bacteria 5 The invention further provides nucleotide sequences coding for the monovalent antigen binding proteins capable of inhibiting the commonly occurring lactococcus bacteriophage P2. The inhibiting binding domains were identified with a high-throughput screening assay, which 10 allows the discrimination of inhibiting from non-inhibiting binding proteins. The involved binding site on phage P2 was characterised by electron microscopy with binding domain fragments conjugated to gold particles. In addition, the cross-reactivity against members of the same family and of other families of bacteriophage was analysed in 15 more detail.
WO 00/65057 PCT/EPOO/03717 - 12 Particular heavy chain variable domains (referred to hereinafter as VHH fragments) of use according to the invention in inhibiting lactococcus bacteriophage P2 comprise the seqences: 5 VHH#1 (SEQ. ID NO. 1) QVQLQESGGG LVQAGGSLRL SCTASRRTGS NWCMGWFRQL AGKEPELVVA LNFDYDMTYY ADSVKGRFTV SRDSGKNTVY LQMNSLKPED TAIYYCAARS GGFSSNRELY DGWGQGTQVT VSS 10 VHH#2 (SEQ. ID NO. 2) QVQLQESGGG LVQAGGSLRL SCTASRRTGS NWSMGWFRQL AGKEREFVVA LNLDYDIPYY ADSVKGRFTV STDSGKNTVY LQMNSLKPED TAIYFCAARS GGFSSNRTYY DYWGQGTQVT VSS 15 VHH#3 (SEQ. ID NO. 3) QVQLQQSGGG LVQRGGSLRL SCTASRRTGS NWSMGWFRQF AGKEPDLLVA LNLDYDVPYY ADSVKGRFTV SGDSGKNTVY LQMNNLKPED TAIYYCAARS GGFSSNRALY DGWGQGTQVT VSS 20 The invention also provides host cells and expression vectors enabling high level production and secretion of the binding proteins. Heavy chain variable domains derived from an immunoglobulin naturally devoid of light chains having a determined antigen specificity may 25 conveniently be obtained by screening expression libraries of cloned fragments of genes encoding Camelid immunoglobulins generated using conventional techniques, as described, for example, in WO 94/04678 and Example 1. Suitable methods to enrich for binding domains recognising the infectious agent, thereby limiting the numbers of clones which 30 have to be screened for the identification of inhibiting fragments are yeast display (WO 94/01567 from Unilever) or phage display. A preferred method to enrich for inhibiting binding domains applicable to the single variable domain binding units described herein, is based 35 on the removal of clones that expose non-inhibiting binding domains, through capture of a complex of the binding domain and the infectious agent of interest by host cells via a receptor protein to which the non-inhibited infectious agent can bind.
WO 00/65057 PCT/EPOO/03717 - 13 Viral infection inhibiting antigen binding proteins according to the invention may be prepared by transforming a host by incorporating a gene encoding the polypeptide as set forth above and expressing said gene in said host. 5 Suitably the host or hosts may be selected from prokarvotic bacteria, such as Gram-negative bacteria, for example E. coli, a-d Gram-positive bacteria, for example B. subtilis and in particular lactic acid bacteria, lower eukaryotes such as yeasts, for example belonging to 10 the genera Saccharomyces, Kluyveromyces, Hansenula or Pichia, or moulds such as those belonging to the genera Aspergillus or Trichoderma. Preferred hosts for use in connection with the present invention are 15 the lower eukaryotic moulds and yeasts, and in particular the lactic acid bacteria, which can be directly used for the fermentation of milk. Techniques for synthesising genes, incorporating them into hosts and 20 expressing genes in hosts are well known in the art and the skilled person would readily be able to put the invention into effect using common general knowledge. Proteins for use according to the invention may be recovered and 25 purified using conventional techniques such as affinity chromatography, ion exchange chromatography or gel filtration chromatography. The binding activity of the binding proteins according to the 30 invention may conveniently be measured by standard techniques known in the art such as enzyme-linked immunoadsorbant assay (ELISA), radioimmune assay (RIA) or by using biosensors. The inhibiting capacity may be examined by the inhibition of plaque formation of phage and viruses, or by a method, which reveals continued cell growth 35 as a measure for resistance against infection. In the case of lactococcus bacteriophage, the high throughput screening assay described in this application, or in an acidification experiment by the fermentation of milk is particularly applicable.
WO 00/65057 PCT/EPOO/03717 - 14 Antigen binding proteins capable of binding to a virus according to the present invention may conveniently be added to food or cosmetic compositions by methods conventional in the art to give products which are protected against infection by the 'particular virus. 5 Alternatively, the antigen binding proteins according to the invention may be formulated into pharmaceutical compositions with pharmaceutically acceptable carriers and/or excipients and optionally other pharmaceutically or cosmetically active ingredients using techniques well known in the art. 10 The following examples are provided by way of illustration only. Techniques used for the manipulation and analysis of nucleic acid materials were performed as described in Sambrook et al, Molecular Cloning, Cold Spring Harbor Press, New York, 2nd Ed. (1989), unless 15 otherwise indicated. Phages were isolated and propagated according to the methods described by Moineau et al, Canadian Journal of Microbiology 38/9, 875-882 (1992). VHH denotes heavy chain variable domain of heavy chain antibodies. 20 Restriction sites are underlined. EXAMPLES 25 Example 1. Induction of a humoral immune response in llama A male llama was immunised with bacteriophage P2 of Lactococcus 2.actis in oil emulsion (1:9 V/V, antigen in water: Specol (Bokhout et al (1981), Immunol. Immunopath., 2, 491-500; Bokhout et al (1986), 30 Infect. Dis., 161-168) subcutaneously and intramuscularly. Per immunisation site 0.75-1.5 ml water in oil emulsion was injected containing 200 ptg phage protein (approx. 6*103 pfu) . Immunisations were performed according to the following time schedule: the second immunisation was performed three weeks after the first injection, and 35 the third immunisation two weeks after the second one. The immune response was followed by titration of serum samples in ELISA with bacteriophage immobilised on Nunc maxi-sorb plates (coat solution 1010 pfu/ml diluted in phosphate buffered saline). After incubation with WO 00/65057 PCT/EPOO/03717 - 15 serum, the bound llama antibodies were detected with polyclonal rabbit-anti-llama antiserum (obtained via immunising rabbits with llama immunoglobulines purified via ProtA and ProtG columns; ID-DLO) and swine-anti-rabbit immunoglobulines (DAKO) conjugated to horse 5 radish peroxidase. Finally the peroxidase enzyme-activity was determined with tetramethylbenzidine and ureaperoxide as substrate and, after termination of the reac ion by adding H:S0 4 , the optical density was measured at 450 nrm. 10 Example 2. Cloning, selection and screening of llama VHH fragments neutralising Lactococcus lactis bacteriophage P2 2.1 Isolation of VHH fragments against Lactoccus lactis bacteriophage P2 15 From the llama, positively responding against bacteriophage P2 as tested in ELISA, a blood sample of about 200 ml was taken and an enriched lymphocyte population was obtained via centrifugation on a Ficoll (Pharmacia) discontinuous gradient. From these cells, total 20 RNA was isolated by guanidium thiocyanate extraction (e.g. via the method described by Chomczynnski and Sacchi (1987), Analytical Biochem., 162, 156-159. After first strand cDNA synthesis using MMLV RT (Gibco-BRL) and random oligonucleotide primers (Pharmacia), DNA fragments encoding VHH fragments and part of the long or short hinge 25 region were amplified by PCR using specific primers: PstI VH - 2B 5'-AGGTSMARCTGCAGSAGTCWGG-3' (SEQ. ID NO. 4) 30 S = C and G, M = A and C, R = A and G, W = A and T, HindIII Lam-07 5'-AACAGTTAAGCTTCCGCTTGCGGCCGCC-GAGCTGGGGTCTTCGCTGTGGTGCG-3' (short hinge) (SEQ. ID NO. 5) 35 HindI II Lam-08 5'-AACAGTTAAGCTTCCGCTTGCGGCCGCTGGTTGTGGTTTTGGTGTCTTGGGTT-3' (long hinge) (SEQ. ID NO. 6) WO 00/65057 PCT/EPOO/03717 - 16 The DNA-fragments generated by PCR were digested with PstI (coinciding with codon 4 and 5 of the VHH dcnain, encoding the amino acids L-Q) and HindIII (introduced at the 5' end of the hinge specific 5 oligonucleotide primers, coinciding with the amino acid sequence S-L T), and cloned in the phagemid vector pUR4676 (identical to pHEN1 (Hoogenboom et al, Nucleic Acids Res. , (1990) , 19, 4133-4137), containing the lacI element as described by Orum et al, Nucleic Acid Res., (1993), 21, 4491-4498) as gene-fragments encoding the 10 VHH-domain including the hinge region fused to the geneIII protein of the E. coli- bacteriophage M13, thereby enabling display of the antibody fragment on the surface of the filamentous phage (McCafferty et al (1990), Nature, 6, 552-554). 15 2.2 Enrichment of lactococcus bacteriophage binding V.H domains via phage display methodology I) A display library with 1x10 7 clones, of which 75% contained a complete VHH encoding insert, was constructed in phagemid vector 20 pUR4676. Phage particles exposing VHH fragments were prepared by infection of E. coli cells harbouring the phagemid with helperphage VCS-M13 (Marks et al (1991), J. Mol. Biol., 222, 581-597). By precipitation of phage from the culture supernatant with PEG6000, free VHH fragments were removed, 25 thereby avoiding a disturbing competition for binding to antigen between phage bound and free V.H domains. II) Phage antibodies binding to lactococcus bacteriophage P2, immobilised on maxisorp immunotubes, were selected from the 30 library via the biopanning procedure (McCafferty et al (1990), Nature, 6, 552-554). After an extensive washing procedure, E. coli phage was eluted from the tube with 0.1 M triethylamine (Baker) by disruption of the antigen-antibody binding with this alkaline shock. After neutralization with 0.5 volume of 1 M 35 Tris-HC1 pH7.4, phage was rescued by transfection into the E. coli host TG1. A renewed selection was performed with phage prepared from the transfected population of E. coli bacteria as WO 00/65057 PCT/EPOO/03717 - 17 was described before. Alternatively, 'in solution' capture of E. coli phage exposing lactococcus phage specific antibody fragments was performed with in vitro biotinylated bacteriophage P2. Antigen-antibody complexes and 5 associated phage particles were pulled out of the solution with streptavidin coated magnetic beads (Dynal) (see Hawkins et al (1992), J. Mol. Biol., 226, 889-896). After washing, E. coli phage was eluted with triethylamine as described before. 10 Individual E. coli clones obtained after the two rounds of selection were grown in wells of microtiter plates, and the production of VHH fragments was induced by the addition of isopropyl-P-D thiogalactopyranoside (IPTG, 0.1 mM). After 16 hours of growth, the culture supernatant of the clones was analysed in ELISA for the 15 presence of VHH fragments, which specifically bind to immobilised bacteriophage P2. Bound VHH fragments were detected with rabbit anti llama VHH polyclonal antibodies followed by incubation with goat anti rabbit polyclonal antibodies conjugated to horse radish peroxidase (BIORAD), or with mouse monoclonal anti-myc antibody followed by 20 incubation with polyclonal rabbit-anti-mouse conjugated to horse radish peroxidase (DAKO). 2.2.1 Alternative enrichment method 25 Following the method of Example 2.2 (I) above, a library of phage bound VHH domains may be prepared. After incubation of the E. coli phage with in vitro biotinylated lactococcus bacteriophage P2 for two hours, E. coli phage clones exposing non-neutralising, bnt phage P2 specific VHH fragments may be captured with an excess of host cells from strain 30 L. lactis. The E. coli phage particles complexed to biotinylated phage P2 via their exposed VHH fragments, but which are not bound to L. lactis (and thereby potentially neutralising), may be captured from solution with virus specific ligands such as streptavidin coated magnetic beads, and thus separated from E. coli phage not bound via 35 their exposed VHH fragment to phage P2. After elution with a pH-shock (0.1 M triethylamine), the phage population enriched for neutralising VHH domains may be rescued by infection of E. coli host cells.
WO 00/65057 PCT/EPOO/03717 - 18 As an alternative method, unlabeled phage P2 can be used instead of biotinylated phage for binding to VHH-exposed E. coli phage. After capture of clones exposing non-neutralising VHH fragments with 5 L. lactis, a population of clones displaying neutralising binding domain proteins can be captured frc- solution with monoclonal or polyclonal antibodies directed against L. lactis bacteriophage P2, which were immobilised on a solid surface or coupled to a matrix. 10 Individual E. coli clones may be grown in wells of microtiter plates, and the production of VHH fragments induced by addition of IPTG (0.1 mM). Culture supernatants containing free VHH domains may be tested in ELISA for binding to L. lactis bacteriophage P2 using the myc-TAG for detection and for their inhibiting capacity in the high throughput 15 assay using the techniques described above. 2.3 Development of a high-throughput screening assay for the identification of bacteriophage neutralising VHH fragments 20 The phage neutralising capacity of the VHH fragments was demonstrated by a continued growth of the host cell L. lactis. As measure for cell growth the acidification in milk was followed with the included pH indicator bromophenol red, which changes from purple-red (pH is 6.5 to 7.0) at the start of cultivation to yellow (pH 4.5 to 5.0) after 8 to 25 15 hours of growth. 50 li1 supernatant of individual clones derived from the selections with E. coli or S. cerevisiae was mixed with 50 pl phage solution (2*109 pfu/ml diluted in semi-skimmed milk supplemented with 0.35% peptone, 0.35% yeast extract, 1% glucose, 0.8% Polymixin B) in a well of a microtiter plate. Subsequently, 100 pl of L. lactis 30 cells (50-fold diluted overnight culture in semi-skimmed milk medium described before, supplemented with 2% bromophenol red) . After 8 to 15 hours of incubation at 30*C, ten neutralising antibodies out of 285 analysed VHH fragments were identified by the change in colour (yellow). Three of these were characterised in detail (see section 35 2.6 and further).
WO 00/65057 PCTIEPOO/03717 - 19 2.4 Sequences of bacteriophage neutralising VHH fragments As indicated in the preceding paragraphs anti-LAB-phage V=H fragments were obtained, which are capable to neutralise lactococcus 5 bacteriophage P2. The sequences of three of such fragments are presented below: VHH#1 (cloned in E. coli phagemid vector pUR3827 and in S. cerevisiae 10 episomal plasmid pUR3834): QVQLQESGGG LVQAGGSLRL SCTASRRTGS NWCMGWFRQL AGKEPELVVA LNFDYDMTYY ADSVKGRFTV SRDSGKNTVY LQMNSLKPED TAIYYCAARS GGFSSNRELY DGWGQGTQVT VSS (SEQ. ID NO. 1) VHH#2 15 (in E. coli plasmid pUR3828 and in S. cerevisiae episomal plasmid pUR3835): QVQLQESGGG LVQAGGSLRL SCTASRRTGS NWSMGWFRQL AGKEREFVVA LNLDYDIPYY ADSVKGRFTV STDSGKNTVY LQMNSLKPED TAIYFCAARS GGFSSNRTYY DYWGQGTQVT VSS (SEQ. ID NO. 2) 20 VHH#3 (in E. coli plasmid pUR3829 and in S. cerevisiae episomal plasmid pUR3836): QVQLQQSGGG LVQRGGSLRL SCTASRRTGS NWSMGWFRQF AGKEPDLLVA LNLDYDVPYY ADSVKGRFTV SGDSGKNTVY LQMNNLKPED TAIYYCAARS GGFSSNRALY DGWGQGTQVT VSS 25 (SEQ. ID NO. 3) Example 3. The efficiency of V.H fragments in neurralising Lactococcus lactis bacteriophage P2 30 3.1 Recloning in episomal plasmid system for production of VsH fragments in S. cerevisiae The VHH encoding genes of clones VHH#1, VHH#2 and VHH#3 were digested with PstI (present at the 5' end of the VHH gene and introduced by 35 primer VH - 2B (SEQ. ID. NO. 1) ) and BstEII (naturally occurring at the 3' end of most VHH genes) and BstEII from the E. coli phagemid vectors pUR3827, pUR3828 and pUR3829 respectively, and cloned WO 00/65057 PCT/EPOO/03717 - 20 in the episomal S. cerevisiae secretion plasmid pUR4547, thereby obtaining pUR3834, pUR3835 and pUR3836 respectively. Plasmid pUR4547 (deposited as CBS100012), with an Ori for autonomous replication in S. cerevisiae, enables the production via the inducible Gal7 promotor; 5 secretion is accomplished by fusing the SUC leader sequence (Harmsen et al (1993), Gene, 125, 115-123) to the amino terminus of the V=H product. The production was examined by analysis of the medium fraction obtained after 48 hours of cultivation at 300C from 5 clones of each construct on a Coomassie blue stained polyacrylamide gel. 10 Plasmid pUR4547 was deposited under the Budapest Treaty at the Centraal Bureau voor Schimmelcultures, Baarn on 18th August 1997 with deposition number: CBS 100012. In accordance with Rule 28(4) EPC, or a similar arrangement from a state not being a contracting state of 15 the EPC, it is hereby requested that a sample of such deposit, when requested, will be submitted to an expert only. 3.2 Construction of stable VyH producing clones of S. cerevisiae by multi-copy integration in the genome 20 Integration of the genes encoding the antibody fragments for establishing stable secreting S. cerevisiae cell lines was accomplished by homologous recombination into the yeast genome. By choosing a multi-copy locus, i.e. the ribosomal DNA-(rDNA) locus 25 containing between 100 and 150 rDNA units, the insertion of multiple copies was forced, thereby yielding high production levels of antibody fragment. The VmH gene of clone #1 was digested with the restriction enzymes SacI (located before the SUC leader sequence) and HindIII (located behind the stopcodon of the VHH gene) and HindIII from 30 the episomal secretion plasmid pUR3834, and cloned in the integration plasmid pUR2778 (Giuseppin et al (1991), WO 91/00920; Driedonks et al (1995), Yeast, 11, 849-864) . This plasmid contains the Gal7 promoter for inducible expression of the VHH gene product (without tags for identification or purification), the selectable markers bla (P 35 lactamase) to discriminate transformants in E. coli by resistance to the antibioticum ampicillin and Leu2d (P-isopropylmalate dehydrogenase) for propagation of transformed S. cerevisiae, an WO 00/65057 PCT/EPOO/03717 - 21 E. coli origin of replication, and finally the flanking homologous sequences for recombination into the genome of S. cerevisiae. Transformants in E. coli container constructs with the V.H gene were identified bv restriction enzvme analysis. Plasmid purified with the 5 Nucleobond AX100 kit was used for transformation of Saccharomyces cerevisiae strain VWK18gall::UPA3 with the lithiumacetate procedure (Gietz and Schiestl (1995), Meth. Mol. Cell. Biol., 5, 255-259). At least 10 individual clones were chosen for production in 50-mi cultures; the medium fraction with the secreted VHH fragments was 10 analysed on a Coommassie blue stained SDS PAGE gel. The clone producing antibody fragment VHH#l most efficiently was coded pUR3858 and it was used for production in a 10-L fermentor. The medium fraction containing the antibody fragment was concentrated by ultrafiltration and further purification was accomplished by means of 15 ion-exchange chromatography (Mono-S-sepharose, Pharmacia). The amount of purified antibody was determined by an OD280 measurement, the micro BCA method, and confirmed by the analysis on a Coomassie stained SDS PAGE gel. 20 3.3 Neutralisation measured by the inhibitory effect on plaque formation To test the neutralising effect of the anti-phage P2 VHH, the reduction in the phage titers was determined. Therefore antibody fragments, 25 produced by S. cerevisiae containing plasmid pUR3834 encoding the neutralising anti-LAB phage VHH#l, or plasmid pUR3831 encoding the LAB phage binding but non-neutralising VHH#4, or construct pUR3850 (PCT/EP98/06991) encoding the neutralising bihead molecule VHH#4-#5, made up of the non-neutralising V.H-fragments VHH#4 and VHH#5 (of the 30 following sequences: VHH#4: (SEQ. ID NO. 7) QVQLQESGGG LVQPGGSLRL SCVVSGEGFS NYPMGWYRQA PGKQRELVAA 35 MSEGGDRTNY ADAVKGRFTI SRDNAKKTVY LQMSSLKPED TAVYYCNAAR WDLGPAPFGS WGQGTQVTVS S WO 00/65057 PCT/EPO0/03717 - 22 VHH#5: (SEQ. ID NO. 8) QVQLQESGGG LVQPGGSLRL SCAVSGAPFR ESTMAWYRQT PGKERETVAF ITSGGSKTYG VSVQGRFTIS RDSDRRTVLL QMNNLQPEDT AVYYCHRALS 5 NTWGQGIQVT VSS were purified as described before. From the monovalent fragments 100 and 5 pg, and 5 and 0.25 pg of the bivalent fragment, were mixed with 5.0*108 phage P2 in 1 ml total volume (diluted in phage buffer: 20 mM 10 Tris-HCl pH 7.4, 100 mM NaCl, 10 mM MgSO 4 ) and incubated for 0.5 hours at 37*C. From this incubation mixture 100 pil undiluted solution, 10-2, 10~4 and 10-6 diluted solution was added to 100 gl of a culture of Lactococcus lactis subsp. cremoris LM0230 (1*109 cfu/ml), which was grown overnight in M17. After the addition of 3 ml of M17 top-agar, 15 the mixture was poured on a plate of M17 containing 0.5% glucose and 10mM CaCl 2 . Plates were incubated overnight at 30*C. Figure 1 shows that at a concentration of 5 pg/ml V H fragment VHH#1 gave a reduction of more than 99% in the phage titre relative to the 20 titre found for the control where no antibody fragment was added to phage. An ELISA positive, lactococcus phage P2 specific V 8 H fragment selected from the same antibody library, which was classified as non neutralising in the high-throughput screening assay, gave no detectable level of neutralization, even at concentrations of 100 25 pg/ml. The bihead molecule VHH#4-#5 did not inhibit infection, at least when the phage P2 was present a such high titres; the example described below shows that the bihead molecule is effective at lower titres of phage. 30 The results demonstrates extremely efficient inhibition (neutralisation) of bacteriophage P2 by the monovale.: fragments here described. 3.4 The efficiency of phage neutralisation determined by the 35 acidification of milk at 30 ml scale In a further aspect, the acidification of milk upon inoculation with WO 00/65057 PCTIEPOO/03717 - 23 lactic acid bacteria at 30 0 C was followed by the regular measurement of the pH. For this purpose 30 ml XVM-glucose medium (skim milk solution containing 0.35% yeast extract, 0.35% peptone and 1% glucose) was inoculated with 300 pl of an overnight culture (10W cfu/ml) of 5 Lactococcus lactis subsp. creroris LM0230. Alternatively, strain C2 was used, which is the LM0230 derived strain producing protease, and therefore these bacteria can grow in skim milk without peptone and glucose. The cultures were incubated for 17 h at 30'C after addition of variable amounts of purified V H fragments. The XVM is acidified by 10 the culture in a period of 8 hours (Figure 2) . When 103 pfu/ml P2 phage was added to the culture of LM0230 or C2 in a parallel experiment, no acidification occurred during the whole period of 17 hours (Figure 2, panel A and B) . Addition of the monovalent antibody fragment VHH#1 (pUR3834) to the culture containing phage P2, resulted 15 in a completely restored acidification profile (Figure 2, panel A). The bihead molecule VHH#4-#5 also prevented phage infection, but the neutralising character stemmed from its bivalency as could be concluded from experiments with the monovalent fragments VHH#4 and 20 VHH#5, which as separately added fragments did not inhibit (Figure 2, panel B). 3.5 Conformation of the monovalent character of the phage neutralising VH fragments 25 In order to exclude possible aggregation of the V H fragments, which might lead to the formation of dimers or higher orders of multimers, as has been observed for single chain antibodies (Holliger et al (1993), Proc. Natl. Acad. Sci., 90, 6444-6448; Kortt et al (1997), 30 Protein Eng., 10, 423-433), the produced molecules were analysed in an ELISA based test. In this assay bacteriophage was immobilised (at a concentration of 1010 pfu per ml of PBS) and as a detection probe in vitro biotinylated bacteriophage was used. Bivalent V. H fragment such as the bihead construct or polyclonal antibody (Figure 3 panel B) 35 present in sera from the immunised llama gave positive responses when incubated with biotinylated phage (prepared with NHS-biotin (Pierce) according to the instruction of the supplier) and horse radish peroxidase labelled streptavidin (DAKO) ; the principle of the assay is WO 00/65057 PCT/EPOO/03717 - 24 shown in Figure 3 (panel A) . In contrast, the non-neutralising VsH fragments VHH#4 and VHH#5 and the neutralising V.H fragments VHH#1, VHH#2 and VHH#3 as well as the polyclonal serum, taken from the llama prior to immunisation with bacteriophage, were not detected with 5 biotinylated phage (Figure 3, panel B). These experiments showed that the produced VHH fragments are monomeric. Therefore the inhibiting effect is not obtained by cross-linking of bacteriophage particles, but rather is determined by the epitope (s) 10 recognised by these particular antibodies. 3.6 Neutralisation of other species of Lactococcus phages Lactococcus phages have been classified into 12 species. Of those 15 only three species have been found to interfere with industrial milk fermentations, i.e. prolate headed c2 species and the isometric-headed 936 (most often found in factories) and P335 species. The bacteriophage P2, which was used for immunisation of the llama and selection of the antibody display library, belongs 20 to the 936 species. Therefore the neutralising capacity of VHH#1 was examined against another member (phage SK1) of the 936 species, but also against two members (phage Q38 and c2) of the prolate headed c2 species. 25 With the microtiter plate assay described in example 2.3 the cross-reactivity and the neutralizing capacity was analysed on the acidification of phage infected cultures as measure for phage resistance. To the mixture of host cells (at a density of 106 cfu/ml) and bacteriophage (at a titer of 103 pfu/ml) variable 30 amounts of antibody fragment VHH#1 were added; after 15 hours of cultivation at 30*C the neutralising activity could be observed by the colour change of the included indicator bromophenol red. The following combinations were tested: L. lactis strain C2 with phage P2, strain SMQ-196 and phage Q38 and bacterial strain LM230 with 35 either phage SKl or phage c2. Besides the described antibody fragment VHH#1 the polyclonal pre- and postimmune sera from the llama were used as negative and positive control as well as not infected bacterial host.
WO 00/65057 PCT/EPOO/03717 - 25 The two tested phages p 2 and SK1 of the isometric-headed 936 species were effectively neutralised by the monovalent antibody fragment (up to a dilution of 47 ng/ml) and post-immune serum (up 5 to a 10-4 fold dilution) . In contrast, the two members Q38 and c2 of the prolate-headed c2 species were not inhibited by VHH#1, not even at concentrations of 0.47 mg/ml, while some neutralisation was observed with post-immune serum at a 10-fold dilution. 10 By using this limited number of different phage types it was concluded that members of the isometric-headed species (used for immunisation) were neutralised effectively by VHH#1, but that phage belonging to the distantly related prolate-headed c2 species were not inhibited. 15 Example 4. Protecting a cheese starter culture from infection with phage during the production and the ripening of semi-hard Gouda cheese 20 Gouda-type cheeses were produced on pilot scale (200 L batches of cheese milk, yielding 4 cheeses of about 6 kg per batch) in open cheese vats. The cheese milk is treated by thermization (68 0 C, 14 sec) and bactofugation, before being standardised to achieve a cheese fat content of about 50% (dry matter). The milk 25 subsequently is pasteurised for 10 sec at 72*C. To the cheese milk, the following components were added: At t = 0 min: bacteriophage 22 in different levels (Table I) At t = 5 min: addition of CaCl 2 (23.1 g/100 1), NaNO 3 (15.5 30 g/100 1) and different levels of the monovalent antibody fragment VHH#1 At t = 10 min: addition of 450g/100 1 starter culture Lactococcus lactis C2 (fully grown in milk) At t = 15 min: addition of 23g/100 1 calf rennet 35 All additions were poured in slowly to the stirred cheese milk to guarantee complete mixing. Further processing (renneting, cutting, curd washing, scalding, draining and filling) was WO 00/65057 PCT/EPOO/03717 - 26 carried out as usual for standard Gouda-type cheese, according to well-known processes as described by Kammerlehner (1) and by De Vries and Van Ginkel (2) . Brining was started when the cheeses had reached a pH of 5.5 to 5.4. Cheeses were ripened at 13 0 C at 3 88% relative humidity. During ripening, samples were taken for the following analysis: " at 2 weeks for the general chemical analysis 1.0 e at 2, 6,13 weeks for protein degradation (total, soluble and amino acid nitrogen (3)) e at 13 weeks for an extensive aroma analysis Samples were stored frozen at -40*C before analysis. Table I Compilation of the 3 separate cheese experiments performed Code Phage Phage in VHH#1 VHH#1 in pH (6 h; 24 (pfu/ml) whey (pg/ml) whey h) 1.1
-
-
- 5.40; 5.22 1.2 -
-
1 + 5.40; 5.21 1.3 1.0 x 10 2.2 x 10 - - 6.00; 5.65 1.4 1.0 x 10' - 1 + 5.46; 5.19 1.5 1.2 x 104 - 1 + 5.40; 5.26 2.1 -
-
-
5.51; 5.16 2.2 1.8 x 10' - 0.1 + 5.59; 5.16 2.3 1.8 x 104 - 1 + 5.57; 5.18 2.4 2.1 x 10' - 0.1 + 5.53; 5.21 2.5 1.7 x 10' 3.0 x 10 - - 5.90; 5.58 3.1 -
-
5.44; 5.22 3.2 1.4 x 10 - 0.1 + 5.45; 5.20 3.3 1.3 x 104 - 1 + 5.49; 5.21 3.4 1.5 x 10 - 1 + 5.48; 5.21 3.5 1.8 x 105 27 0.1 5.45; 5.24 20 WO 00/65057 PCTIEPOO/03717 - 27 The results depicted in Table I clearly show that addition of the monovalent antibody fragment VHH#1 to the 22 phage infected cheese milk, prior to the addition of the starter culture L. lactis C2, protects the cul-ure against phage infection. Phages 5 are not detected anymore in the whey even if 'the cheese milk is infected with 1.5 x 105 pfu/mi, when 1 pg/ml of VHH#1 is present. The acidification of the cheese milk is as expected (cf. 3.1 with 3.4) . When the antibody fragment is not added, phages are detected in the whey and the acidification slows down 10 significantly (cf. 1.1 and 1.3 or 2.1 and 2.5). When the antibody fragment is added, the activity still can be found back in the whey, even if the phage is added prior to the antibody. This proves that the antibody is present in excess and is able to neutralise the phage completely. As can be seen from experiment 15 3.5, the limit is possibly reached when 0.1 pg/ml VHH#1 is added to cheese milk infected with 1.8 x 105 pfu/ml. This level of phage infection is considered to be extremely high in a normal operating cheese plant. 20 To determine if the neutralisation of the phage during the initial cheese fermentation is enough to obtain a normal cheese ripening process, the chemical composition of the cheeses has been determined after 2 weeks as well as the proteolysis after 2, 6 and 13 weeks (Table II and III). 25 Table II Chemical composition cheeses after 2 weeks Analys %Humidity %Fat %Fat %Salt %Salt pH is i.d.s. i.d.s. 1.1 41.4 29.6 50.4 1.9 3.1 5.17 1.2 41.6 29.4 50.3 1.9 3.2 5.16 1.3 40.9 29.9 50.4 2.3 3.9 5.63 1.4 40.1 29.6 50.2 1.9 j3.3 5.18 1.5 41.5 29.9 51.0 1.8 - 3.1 5.14 2.1 42.5 29.2 50.9 2.2 3.8 5.13 2.2 41.2 29.7 50.6 2.2 3.7 5.14 2.3 41.2 30.0 51.0 2.2 3.7 5.15 WO 00/65057 PCT/EP00/03717 - 28 2.4 39.7 30.9 51.0 1.8 3.0 5.19 2.5 39.6 31.2 51.4 1.8 3.0 5.60 3.1 42.3 30.0 51.7 1.7 3.0 5.16 3.2 41.7 30.3 51.8 1.7 2.9 5.17 3.3 41.4 30.2 51.4 1.6 2.8 5.19 3.4 40.4 30.5 51.0 1.5 2.6 5.22 3.5 40.4 31.1 51.8 1.8 2.8 5.26 Table III Proteolysis during ripening of the cheeses (Soluble Nitrogen/Total Nitrogen; Amino acid Nitrogen/TN; AN/SN) 5 SN/T AN/T AN/SN N N Weeks 2 6 13 2 6 13 2 6 13 1.1 7.70 11.80 17.19 1.39 2.13 2.78 18.06 18.08 16.19 1.2 7.86 12.31 17.47 1.53 2.23 2.90 19.50 18.14 16.62 1.3 7.36 11.33 15.33 2.61 4.25 5.79 35.42 37.53 37.76 1.4 7.60 12.34 19.32 2.15 2.47 3.39 28.32 20.0 17.55 1.5 7.81 12.94 17.76 1.63 2.65 4.11 20.92 20.48 23.17 2.1 6.77 10.71 14.96 1.94 2.02 2.75 28.69 18.87 18.39 2.2 6.75 11.48 16.14 1.65 2.10 3.37 24.48 18.33 20.90 2.3 6.11 11.95 16.72 1.84 2.93 3.83 30.13 24.50 22.90 2.4 6.72 11.42 17.15 2.00 2.28 3.60 29.77 19.96 20.98 2.5 7.53 12.01 16.92 2.93 4.30 6.16 38.87 35.76 36.44 3.1 8.33 12.49 17.28 1.13 1.86 2.65 13.51 14.86 15.36 3.2 7.72 12.60 17.51 1.33 2.09 3.10 17.27 16.63 17.68 3.3 8.25 12.33 17.75 1.47 2.22 3.81 17.78 18.05 21.05 3.4 8.21 12.90 18.95 1.58 2.75 4.42 19.20 21.32 23.33 3.5 7.96 12.39 18.34 1.49 2.59 4.34 18.77 20.93 23.68 From the data in Table II it can be concluded that the chemical composition of a cheese is not much influenced by the presence of 10 a phage apart from the acidification (cf. 1.3 and 2.5 with the other data). Consequently an additional ripening period of 2 weeks, does not restore the acidification capacity of the starter WO 00/65057 PCT/EPOO/03717 - 29 culture upon infection with the phage, unless the neutralising antibody VHH#l is added prior to the starter culture. The data from Table III show that proteolysis, that is one of the major indicative parameters for cheese ripening, is abnormal in phage 5 infected cheeses and that this once again can be normalised if VHH#1 is added prior to the addition of the starter culture (cf. The AN/TN and AN/SN data of 1.3 and 2.5 with the other data). Too many amino acids are liberated when the phage is not neutralised by VHH#1, indicating an unbalanced proteolysis and therefore and 10 off-flavoured cheese. References 1. Kammerlehrer, J. (1989) Lab-Kase Technologie. Band III, p 15 642-643. In Molkereitechnik Band 84/85. Verlag Th. Mann, Gelsenkircher-Buer, ISBN 3-7862-0083-1 2. De. Vries E. and van Ginkel, W. (1980) Test of a curd-making tank. Type "Damrow Double 0" with a capacity of 16000 L, manufactured by DEC. NIZO Rapport R113 20 3. Noomen, A., (1977) Noordhollandse Meshanger Cheese: a model for research on cheese ripening.2. The ripening of the cheese. Neth. Milk Diary J. 31, 75-102

Claims (16)

1. A method of inhibiting viral infection using a monovalent 5 antigen binding protein comprising a single variable domain binding unit, or a functional equivalent thereof, capable of binding to a virus.
2. A method according to claim 1 wherein the single domain variable 10 domain binding unit comprises a heavy chain variable domain derived from an immunoglobulin devoid of light chains, or a functional equivalent thereof.
3. A method according to claim 1 or claim 2 wherein the single 15 variable domain binding unit comprises a heavy chain variable domain derived from a Camelid irmunoglobulin or a functional equivalent thereof.
4. A method according to any one of claims 1 to 3 wherein the virus 20 is a lactococcal bacteriophage.
5. A method according to any one of claims 1 to 3 wherein the virus is pathogenic to humans or animals. 25
6. A method according to any one of claims 1 to 4 wherein the virus is pathogenic to plants.
7. Use of a monovalent antigen binding protein comprising a single variable domain binding unit, or a functional equivalent 30 thereof, capable of binding to a virus in inhibiting viral infection.
8. Use of a monovalent antigen binding protein comprising a single variable domain binding unit, or a functional equivalent 35 thereof, capable of binding to a virus in the preparation of a medicament for inhibiting viral infection. WO 00/65057 PCT/EPOO/03717 - 31
9. A food product comprising a single variable domain binding unit, or a functional equivalent thereof, capable of binding to a virus. 5
10. A pharmaceutical or cosmetic composition comprising a single variable domain binding unit, or a functional equivalent thereof, capable of binding to a virus.
11. A monovalent antigen binding protein capable of inhibiting viral 10 infection comprising a heavy chain variable domain comprising an amino acid sequence as shown in SEQ. ID No. 1, 2 or 3.
12. Nucleotide sequences coding for a protein according to claim 11. 15
13. Expression vector comprising a nucleotide sequence according to claim 12.
14. A host cell transformed with a vector according to claim 13. 20
15. Method for selecting an antigen binding protein capable of inhibiting viral infection of a host cell comprising the steps of: i) complexing an antigen binding protein with a target virus, 25 ii) exposing the antigen binding protein-virus complex of step (i) to an excess of host cells, iii) removing the host cells and any associated antigen binding 30 protein-virus complex, iv) capturing antigen binding protein-virus complex not taken up by the host cells in step (ii) with virus specific ligands to separate virus specific antigen binding proteins 35 from non-binding proteins. WO 00/65057 PCT/EPOO/03717 - 32
16. Method for identifying an antigen binding protein capable of inhibiting bacteriophage infection of a lactic acid bacterial cell host comprising the sreps of: 5 i) culturing of bacterial host cells in the presence of antigen binding prczein and bacteriophage, ii) assaying said culture for active cell growth manifest in a change in pH of the culture growth medium. 10
AU41202/00A 1999-04-22 2000-04-19 Inhibition of viral infection using monovalent antigen-binding proteins Ceased AU776824B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP99303117 1999-04-22
EP99303117 1999-04-22
PCT/EP2000/003717 WO2000065057A1 (en) 1999-04-22 2000-04-19 Inhibition of viral infection using monovalent antigen-binding proteins

Publications (2)

Publication Number Publication Date
AU4120200A true AU4120200A (en) 2000-11-10
AU776824B2 AU776824B2 (en) 2004-09-23

Family

ID=8241341

Family Applications (1)

Application Number Title Priority Date Filing Date
AU41202/00A Ceased AU776824B2 (en) 1999-04-22 2000-04-19 Inhibition of viral infection using monovalent antigen-binding proteins

Country Status (11)

Country Link
US (2) US6528056B1 (en)
EP (1) EP1169453A1 (en)
CN (2) CN1252264C (en)
AP (1) AP1447A (en)
AU (1) AU776824B2 (en)
BR (1) BR0009866A (en)
CA (1) CA2370351A1 (en)
ID (1) ID30380A (en)
OA (1) OA11862A (en)
WO (1) WO2000065057A1 (en)
ZA (1) ZA200107822B (en)

Families Citing this family (135)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4731907B2 (en) * 2002-09-16 2011-07-27 アドバンスド バイオニュートリション コーポレーション Protein and peptide expression for passive immunity
FR2846667B1 (en) * 2002-11-06 2004-12-31 Pasteur Institut VARIABLE FRAGMENTS OF SINGLE-CHAIN CAMELIDE ANTIBODIES DIRECTED AGAINST BETA-AMYLOID PEPTIDE 1-42 AND THEIR APPLICATIONS FOR DIAGNOSIS AND TREATMENT OF NEUROAGREGATIVE DISEASES
US20060034845A1 (en) 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
AU2003286004A1 (en) 2002-11-08 2004-06-07 Ablynx N.V. Single domain antibodies directed against interferon- gamma and uses therefor
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
WO2004041865A2 (en) 2002-11-08 2004-05-21 Ablynx N.V. Stabilized single domain antibodies
EP2368554B1 (en) 2003-04-08 2014-12-24 Progenics Pharmaceuticals, Inc. Pharmaceutical formulations containing methylnaltrexone
US8105592B2 (en) 2004-11-25 2012-01-31 Vhsquared Limited Heavy chain and single domain antibodies
EP2365000A3 (en) 2005-05-18 2013-01-16 Ablynx N.V. Improved nanobodiesTM against tumor necrosis factor-alpha
PL3415535T3 (en) 2005-05-20 2021-06-14 Ablynx N.V. Improved nanobodies tm for the treatment of aggregation-mediated disorders
GB0522460D0 (en) * 2005-11-03 2005-12-14 Prendergast Patrick T Composition and method for the treatment of avian influenza
JP2010505946A (en) 2006-10-10 2010-02-25 アカデミシュ ジーケンハウス ビイ デ ユニヴェアズィテート ファン アムステルダム Complement inhibition to improve nerve regeneration
EP2557090A3 (en) 2006-12-19 2013-05-29 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
WO2008074840A2 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders
EP2125024B1 (en) 2007-03-23 2013-02-13 TO-BBB Holding B.V. Targeted intracellular delivery of antiviral agents
EP3263571B2 (en) 2007-03-29 2023-08-23 Progenics Pharmaceuticals, Inc. Crystal form of (r)-n-methylnaltrexone bromide and uses thereof
WO2009004065A2 (en) 2007-07-03 2009-01-08 Ablynx N.V. Providing improved immunoglobulin sequences by mutating cdr and/or fr positions
AU2008328781A1 (en) 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
CA2713568C (en) 2008-02-06 2016-09-20 Progenics Pharmaceuticals, Inc. Preparation and use of (r),(r)-2,2'-bis-methylnaltrexone
DE112009000507T5 (en) 2008-03-05 2011-02-10 Ablynx Nv Novel antigen-binding dimer complexes, process for their preparation and their use
EP2947097A1 (en) 2008-04-07 2015-11-25 Ablynx N.V. Amino acid sequences directed against the Notch pathways and uses thereof
CN102089325A (en) 2008-04-17 2011-06-08 埃博灵克斯股份有限公司 Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
CA2720210A1 (en) * 2008-04-18 2009-10-22 Unilever Plc Compositions comprising antibodies or antibody fragments
ES2713864T3 (en) * 2008-06-05 2019-05-24 Ablynx Nv Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US20100068185A1 (en) * 2008-09-17 2010-03-18 Omnilytics, Incorporated Methods for introducing bacteriophage into the interiors of quantities of edible agricultural products and edible agricultural products with phage-containing preparations in the interiors thereof
US10005830B2 (en) 2009-03-05 2018-06-26 Ablynx N.V. Antigen binding dimer-complexes, methods of making/avoiding and uses thereof
CN101846687A (en) * 2009-03-25 2010-09-29 中国科学院大连化学物理研究所 Protein equalizer with immobilized scFv library, preparation method and application
PT2424889E (en) 2009-04-30 2015-11-12 Ablynx Nv Method for the production of domain antibodies
HUE035773T2 (en) 2009-06-05 2018-05-28 Ablynx Nv Trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections
WO2011003622A1 (en) 2009-07-10 2011-01-13 Ablynx N.V. Method for the production of variable domains
LT2805731T (en) 2009-09-03 2019-02-11 Ablynx N.V. Stable formulations of polypeptides and uses thereof
KR20110048672A (en) * 2009-11-03 2011-05-12 엘지전자 주식회사 Air cleaning filter comprising kimchi lactic acid bacteria and anticeptic, and process for producing the same
EP2507262A1 (en) 2009-11-30 2012-10-10 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
WO2011083141A2 (en) 2010-01-08 2011-07-14 Ablynx Nv Method for generation of immunoglobulin sequences by using lipoprotein particles
CA2788993A1 (en) 2010-02-05 2011-08-11 Ablynx N.V. Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
PT2533761T (en) 2010-02-11 2019-06-17 Ablynx Nv Methods and compositions for the preparation of aerosols
US9556273B2 (en) 2010-03-29 2017-01-31 Vib Vzw Anti-macrophage mannose receptor single variable domains for targeting and in vivo imaging of tumor-associated macrophages
US9101674B2 (en) 2010-03-29 2015-08-11 Vib Vzw Targeting and in vivo imaging of tumor-associated macrophages
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
US20130261288A1 (en) 2010-10-29 2013-10-03 Ablynx N.V. Method for the production of immunoglobulin single variable domains
PT2691415T (en) 2011-03-28 2018-10-19 Ablynx Nv Method for producing solid formulations comprising immunoglobulin single variable domains
WO2012152823A1 (en) 2011-05-09 2012-11-15 Ablynx Nv Method for the production of immunoglobulin single variable domains
EP2714736A1 (en) 2011-05-27 2014-04-09 Ablynx N.V. Inhibition of bone resorption with rankl binding peptides
EP2723772A1 (en) 2011-06-23 2014-04-30 Ablynx N.V. Immunoglobulin single variable domains directed against ige
WO2013041722A1 (en) 2011-09-23 2013-03-28 Ablynx Nv Prolonged inhibition of interleukin-6 mediated signaling
EP2855526B1 (en) 2012-05-24 2018-11-07 VIB vzw Anti-macrophage mannose receptor immunoglobulin single variable domains for targeting and in vivo imaging of tumor-associated macrophages
WO2014087010A1 (en) 2012-12-07 2014-06-12 Ablynx N.V. IMPROVED POLYPEPTIDES DIRECTED AGAINST IgE
AU2014211355B2 (en) 2013-01-30 2017-07-13 Vib Vzw Novel chimeric polypeptides for screening and drug discovery purposes
PT2953973T (en) 2013-02-05 2019-10-25 Univ Brussel Vrije Muscarinic acetylcholine receptor binding agents and uses thereof
EP2968487B1 (en) 2013-03-15 2019-01-30 Vib Vzw Anti-macrophage mannose receptor single variable domains for use in cardiovascular diseases
AU2014273418B2 (en) 2013-04-29 2020-04-23 Biotalys NV Agrochemical compositions comprising antibodies binding to sphingolipids
NL1040254C2 (en) 2013-05-17 2014-11-24 Ablynx Nv Stable formulations of immunoglobulin single variable domains and uses thereof.
EP2883883A1 (en) 2013-12-16 2015-06-17 Cardio3 Biosciences S.A. Therapeutic targets and agents useful in treating ischemia reperfusion injury
WO2015121092A1 (en) 2014-01-30 2015-08-20 Vib Vzw Opioid receptor binding agents and uses thereof
NL2013661B1 (en) 2014-10-21 2016-10-05 Ablynx Nv KV1.3 Binding immunoglobulins.
WO2016012363A1 (en) 2014-07-22 2016-01-28 Vib Vzw Methods to select for agents that stabilize protein complexes
EP3180037A1 (en) 2014-07-29 2017-06-21 Vrije Universiteit Brussel Radio-labelled antibody fragments for use in the prognosis, diagnosis of cancer as well as for the prediction of cancer therapy response
MX356502B (en) 2014-07-29 2018-05-31 Univ Brussel Vrije Radio-labelled antibody fragments for use in the prevention and/or treatment of cancer.
US10858666B2 (en) 2014-11-05 2020-12-08 Biotalys Transgenic plants expressing a variable domain of a heavy chain antibody (VHH) that binds to a sphingolipid of a fungus
US11426468B2 (en) 2014-12-19 2022-08-30 Ablynx N.V. Cysteine linked nanobody dimers
CN104845937B (en) * 2015-04-09 2017-12-29 浙江大学 Secrete anti-WYMV monoclonal antibody hybridoma cell strain and its monoclonal antibody application
BR112018000672A2 (en) 2015-07-17 2018-09-18 Univ Brussel Vrije radiolabelled antibody fragments for use in cancer treatment
JP7256011B2 (en) 2015-11-27 2023-04-11 アブリンクス エン.ヴェー. Polypeptides that inhibit CD40L
WO2017182603A1 (en) 2016-04-22 2017-10-26 Université Libre de Bruxelles A new biomarker expressed in pancreatic beta cells useful in imaging or targeting beta cells
WO2017182605A1 (en) 2016-04-22 2017-10-26 Université Libre de Bruxelles A new biomarker expressed in pancreatic beta cells useful in imaging or targeting beta cells
AU2017259876A1 (en) 2016-05-02 2018-10-25 Ablynx Nv Treatment of RSV infection
WO2018007442A1 (en) 2016-07-06 2018-01-11 Ablynx N.V. Treatment of il-6r related diseases
WO2018029182A1 (en) 2016-08-08 2018-02-15 Ablynx N.V. Il-6r single variable domain antibodies for treatment of il-6r related diseases
EP3512880A1 (en) 2016-09-15 2019-07-24 Ablynx NV Immunoglobulin single variable domains directed against macrophage migration inhibitory factor
KR102665596B1 (en) 2016-11-16 2024-05-14 아블린쓰 엔.브이. T cell recruiting polypeptide capable of binding CD123 and TCR alpha/beta
WO2018099968A1 (en) 2016-11-29 2018-06-07 Ablynx N.V. Treatment of infection by respiratory syncytial virus (rsv)
WO2018158335A1 (en) 2017-02-28 2018-09-07 Vib Vzw Means and methods for oral protein delivery
US20200033347A1 (en) 2017-04-18 2020-01-30 Universite Libre De Bruxelles Biomarkers And Targets For Proliferative Diseases
EP3621990A1 (en) 2017-05-11 2020-03-18 VIB vzw Glycosylation of variable immunoglobulin domains
BR112019025392A2 (en) 2017-06-02 2020-07-07 Ablynx N.V. aggrecan-binding immunoglobulins
AU2018277102A1 (en) 2017-06-02 2019-12-19 Ablynx N.V. Polypeptides binding ADAMTS5, MMP13 and Aggrecan
MX2019014400A (en) 2017-06-02 2020-02-10 Merck Patent Gmbh Adamts binding immunoglobulins.
US20200190216A1 (en) 2017-06-02 2020-06-18 Merck Patent Gmbh Mmp13 binding immunoglobulins
WO2019016237A1 (en) 2017-07-19 2019-01-24 Vib Vzw Serum albumin binding agents
WO2019086548A1 (en) 2017-10-31 2019-05-09 Vib Vzw Novel antigen-binding chimeric proteins and methods and uses thereof
WO2019155041A1 (en) 2018-02-12 2019-08-15 Vib Vzw Gβγ COMPLEX ANTIBODIES AND USES THEREOF
US11858960B2 (en) 2018-03-01 2024-01-02 Vrije Universiteit Brussel Human PD-L1-binding immunoglobulins
CN116942851A (en) 2018-03-23 2023-10-27 布鲁塞尔自由大学 Wnt signaling agonist molecules
MX2020010091A (en) 2018-03-27 2021-01-15 Umc Utrecht Holding Bv Targeted thrombolysis for treatment of microvascular thrombosis.
CA3138028A1 (en) 2019-04-29 2020-11-05 Confo Therapeutics N.V. Chimeric proteins and methods to screen for compounds and ligands binding to gpcrs
US20220289837A1 (en) 2019-04-30 2022-09-15 Vib Vzw Cystic Fibrosis Transmembrane Conductance Regulator Stabilizing Agents
EP3976650A1 (en) 2019-05-28 2022-04-06 Vib Vzw Cancer treatment by targeting plexins in the immune compartment
WO2020239934A1 (en) 2019-05-28 2020-12-03 Vib Vzw Cd8+ t-cells lacking plexins and their application in cancer treatment
EP4048703A1 (en) 2019-10-21 2022-08-31 Vib Vzw Nanodisc-specific antigen-binding chimeric proteins
WO2021095031A2 (en) 2019-11-11 2021-05-20 Ibi-Ag Innovative Bio Insecticides Ltd. Insect control nanobodies and uses thereof
US20220411495A1 (en) 2019-11-27 2022-12-29 Vib Vzw Positive allosteric modulators of the calcium-sensing receptor
GB201918279D0 (en) 2019-12-12 2020-01-29 Vib Vzw Glycosylated single chain immunoglobulin domains
JP2023506961A (en) 2019-12-20 2023-02-20 フエー・イー・ベー・フエー・ゼツト・ウエー Nanobody exchange chromatography
WO2021140205A1 (en) 2020-01-10 2021-07-15 Confo Therapeutics N.V. Methods for generating antibodies and antibody fragments and libraries comprising same
WO2021156490A2 (en) 2020-02-06 2021-08-12 Vib Vzw Corona virus binders
CA3173090A1 (en) 2020-02-25 2021-09-02 Vib Vzw Leucine-rich repeat kinase 2 allosteric modulators
JP2023519975A (en) 2020-03-31 2023-05-15 バイオタリス・エン・フェー antifungal polypeptide
WO2021213435A1 (en) 2020-04-22 2021-10-28 迈威(上海)生物科技股份有限公司 Single variable domain antibody targeting human programmed death ligand 1 (pd-l1) and derivative thereof
WO2021229104A1 (en) 2020-05-15 2021-11-18 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
WO2022003156A1 (en) 2020-07-02 2022-01-06 Oncurious Nv Ccr8 non-blocking binders
US20230265478A1 (en) 2020-07-31 2023-08-24 Biotalys NV Methods of increasing recombinant protein yields
WO2022063957A1 (en) 2020-09-24 2022-03-31 Vib Vzw Biomarker for anti-tumor therapy
CA3196737A1 (en) 2020-09-24 2022-03-31 Massimiliano Mazzone Combination of p2y6 inhibitors and immune checkpoint inhibitors
AU2021350156A1 (en) 2020-09-25 2023-06-08 Ablynx Nv Polypeptides comprising immunoglobulin single variable domains targeting il-13 and ox40l
WO2022117569A1 (en) 2020-12-02 2022-06-09 Oncurious Nv A ccr8 antagonist antibody in combination with a lymphotoxin beta receptor agonist antibody in therapy against cancer
US20240018248A1 (en) 2020-12-02 2024-01-18 Vib Vzw An ltbr agonist in combination therapy against cancer
US11897951B2 (en) 2020-12-18 2024-02-13 Ablynx N.V. Polypeptides comprising immunoglobulin single variable domains targeting IL-6 and TNF-α
GB202020502D0 (en) 2020-12-23 2021-02-03 Vib Vzw Antibody composistion for treatment of corona virus infection
EP4267621A1 (en) 2020-12-24 2023-11-01 Vib Vzw Murine cross-reactive human ccr8 binders
EP4267617A1 (en) 2020-12-24 2023-11-01 Vib Vzw Human ccr8 binders
CA3206124A1 (en) 2020-12-24 2022-06-30 Vib Vzw Non-blocking human ccr8 binders
CN117794566A (en) 2021-02-05 2024-03-29 Vib研究所 Sha Bei viral binding agents
JP2024506020A (en) 2021-02-05 2024-02-08 ブイアイビー ブイゼットダブリュ Sarbecovirus binder
CA3211257A1 (en) 2021-02-17 2022-08-25 Vib Vzw Inhibition of slc4a4 in the treatment of cancer
CA3211270A1 (en) 2021-02-19 2022-08-25 Vib Vzw Cation-independent mannose-6-phosphate receptor binders
WO2022199804A1 (en) 2021-03-24 2022-09-29 Vib Vzw Nek6 inhibition to treat als and ftd
WO2022242892A1 (en) 2021-05-17 2022-11-24 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
WO2022268993A1 (en) 2021-06-23 2022-12-29 Vib Vzw Means and methods for selection of specific binders
EP4365199A1 (en) 2021-06-29 2024-05-08 Shandong Simcere Biopharmaceutical Co., Ltd. Cd16 antibody and use thereof
CN117751143A (en) 2021-07-30 2024-03-22 山东先声生物制药有限公司 anti-PVRIG/anti-TIGIT bispecific antibodies and uses
WO2023016828A2 (en) 2021-07-30 2023-02-16 Vib Vzw Cation-independent mannose-6-phosphate receptor binders for targeted protein degradation
WO2023057601A1 (en) 2021-10-06 2023-04-13 Biotalys NV Anti-fungal polypeptides
WO2023098846A1 (en) 2021-12-03 2023-06-08 江苏先声药业有限公司 Anti-bcma nanobody and use thereof
TW202342508A (en) 2021-12-17 2023-11-01 比利時商艾伯霖克斯公司 POLYPEPTIDES COMPRISING IMMUNOGLOBULIN SINGLE VARIABLE DOMAINS TARGETING TCRαβ, CD33 and CD123
WO2023135198A1 (en) 2022-01-12 2023-07-20 Vib Vzw Human ntcp binders for therapeutic use and liver-specific targeted delivery
WO2023148291A1 (en) 2022-02-02 2023-08-10 Biotalys NV Methods for genome editing
WO2023148397A1 (en) 2022-02-07 2023-08-10 Vib Vzw Engineered stabilizing aglycosylated fc-regions
WO2023198848A1 (en) 2022-04-13 2023-10-19 Vib Vzw An ltbr agonist in combination therapy against cancer
WO2023213751A1 (en) 2022-05-02 2023-11-09 Umc Utrecht Holding B.V Single domain antibodies for the detection of plasmin-cleaved vwf
WO2023222825A1 (en) 2022-05-18 2023-11-23 Vib Vzw Sarbecovirus spike s2 subunit binders
WO2024008755A1 (en) 2022-07-04 2024-01-11 Vib Vzw Blood-cerebrospinal fluid barrier crossing antibodies
WO2024068744A1 (en) 2022-09-27 2024-04-04 Vib Vzw Antivirals against human parainfluenza virus
WO2024105091A1 (en) 2022-11-15 2024-05-23 Imec Vzw Method and system for droplet manipulation
US20240200085A1 (en) 2022-12-15 2024-06-20 Aarhus Universitet Synthetic activation of multimeric transmembrane receptors
WO2024133937A1 (en) 2022-12-22 2024-06-27 Biotalys NV Methods for genome editing

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2590675B1 (en) * 1985-11-27 1990-10-12 Transia MIXTURE OF SPECIFIC ANTIBODIES OF LACTIC BACTERIOPHAGES AND ITS APPLICATIONS TO THE DETECTION AND NEUTRALIZATION OF SAID BACTERIOPHAGES
GB8927230D0 (en) * 1989-12-01 1990-01-31 Unilever Plc Reagents
WO1994004678A1 (en) * 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulins devoid of light chains
ES2162863T3 (en) * 1993-04-29 2002-01-16 Unilever Nv PRODUCTION OF ANTIBODIES OR FRAGMENTS (FUNCTIONALIZED) OF THE SAME DERIVED FROM HEAVY CHAIN IMMUNOGLOBULINS OF CAMELIDAE.
CN1203178C (en) * 1997-10-27 2005-05-25 尤尼利弗公司 Multivalent antigen-binding proteins
EP0954978B1 (en) * 1998-03-12 2011-11-30 VHsquared Limited New products comprising inactivated yeasts or moulds provided with active antibodies

Also Published As

Publication number Publication date
OA11862A (en) 2006-03-02
CN1891715A (en) 2007-01-10
US6528056B1 (en) 2003-03-04
AP1447A (en) 2005-08-12
BR0009866A (en) 2002-01-08
CN1252264C (en) 2006-04-19
CA2370351A1 (en) 2000-11-02
US7132121B2 (en) 2006-11-07
AU776824B2 (en) 2004-09-23
CN1351662A (en) 2002-05-29
EP1169453A1 (en) 2002-01-09
ZA200107822B (en) 2002-09-23
AP2001002301A0 (en) 2001-12-31
ID30380A (en) 2001-11-29
US20030165511A1 (en) 2003-09-04
WO2000065057A1 (en) 2000-11-02
CN100434441C (en) 2008-11-19

Similar Documents

Publication Publication Date Title
US6528056B1 (en) Inhibition of viral infection using antigen-binding proteins
De Haard et al. Llama antibodies against a lactococcal protein located at the tip of the phage tail prevent phage infection
Harmsen et al. Selection and optimization of proteolytically stable llama single-domain antibody fragments for oral immunotherapy
Abbady et al. Evaluation of a nanobody phage display library constructed from a Brucella-immunised camel
JP4656478B2 (en) Antibody library
CN111848789B (en) Single chain antibody for resisting SARS-COV-2 virus S protein and its use
US6818748B2 (en) Cloning, expression, sequencing, and functional enhancement of monoclonal ScFv antibody against Venezuelan equine encephalitis virus (VEE)
Chackerian et al. Peptide epitope identification by affinity selection on bacteriophage MS2 virus-like particles
CN111057145B (en) Porcine reproductive and respiratory syndrome virus Nsp2 protein nano antibody and application thereof
CN110526967A (en) A kind of staphylococcus aureus toxin A nano antibody A13, application and kit
Daoudi et al. Production and characterization of anti-nisin Z monoclonal antibodies: suitability for distinguishing active from inactive forms through a competitive enzyme immunoassay
Sapats et al. Generation of chicken single chain antibody variable fragments (scFv) that differentiate and neutralize infectious bursal disease virus (IBDV)
Tesar et al. Monoclonal antibody against pIII of filamentous phage: an immunological tool to study pIII fusion protein expression in phage display systems
CN106632670B (en) The single-chain antibody and preparation method thereof of one boar source property anti-swine infectious enterogastritis virus
Sepulveda et al. Design and testing of PCR primers for the construction of scFv libraries representing the immunoglobulin repertoire of rats
CN105504053B (en) A kind of Porcine epidemic diarrhea virus M protein-specific heavy chain antibody
Shahmirzaie et al. Generation and molecular docking analysis of specific single-chain variable fragments selected by phage display against the recombinant nucleocapsid protein of fig mosaic virus
CN105859879B (en) A kind of single-chain antibody of Antifish lymphocystis virus
CN105481979B (en) Human source anti-rabies virus glycoprotein neutrality antibody RV3A5 and its application
CN112608383B (en) Single-chain antibody for resisting paralichthys rhabdovirus
CN113527477B (en) Swine-derived anti-PDCoV-N protein scFv, expression vector, construction method and application thereof
Tout et al. Phage display and bacterial expression of a recombinant Fab specific for Pseudomonas aeruginosa serotype O6 lipopolysaccharide
Chevigné et al. Use of bifunctional hybrid β-lactamases for epitope mapping and immunoassay development
Naylor Development of a Dusky kob scFv gene phage display library for the discovery of antibodies to Brome mosaic virus-a proxy for a novel, emerging fish pathogen
Raynes Structural and Immunological analysis of the T antigen: a vaccine candidate for Streptococcus pyogenes